3,424
Views
9
CrossRef citations to date
0
Altmetric
Review

Alternative splicing and MicroRNA: epigenetic mystique in male reproduction

, , , &
Pages 162-175 | Received 13 Jul 2021, Accepted 26 Dec 2021, Published online: 22 Jan 2022

References

  • Tournaye H, Krausz C, Oates RD. Novel concepts in the aetiology of male reproductive impairment. Lancet Diabetes Endocrinol. 2017;5(7):544–553.
  • Oatley JM, Brinster RL. Regulation of spermatogonial stem cell self-renewal in mammals. Annu Rev Cell Dev Biol. 2008;24:263–286.
  • Yan W. Male infertility caused by spermiogenic defects: lessons from gene knockouts. Mol Cell Endocrinol. 2009;306:24–32.
  • Zhang Y, Shi J, Rassoulzadegan M, et al. Sperm RNA code programmes the metabolic health of offspring. Nat Rev Endocrinol. 2019;15:489–498.
  • Braunschweig U, Gueroussov S, Plocik AM, et al. Dynamic integration of splicing within gene regulatory pathways. Cell. 2013;152:1252–1269.
  • Merkin J, Russell C, Chen P, et al. Evolutionary dynamics of gene and isoform regulation in mammalian tissues. Science. 2012;338:1593–1599.
  • Qing G, Lu Q, Xiong Y, et al. New opportunities and challenges of smart polymers in post-translational modification proteomics. Adv Mater. 2017;29:20.
  • Nilsen TW, Graveley BR. Expansion of eukaryotic proteome by alternative splicing. Nature. 2010;463:457–463.
  • Scotti MM, Swanson MS. RNA-mis-splicing in disease. Nat Rev Genet. 2016;17:19–32.
  • Lee Y, Rio DC. Mechanisms and regulation of alternative pre-mRNA Splicing. Annu Rev Biochem. 2015;84:291–323.
  • Fica SM, Tuttle N, Novak T, et al. RNA catalyses nuclear pre-mRNA splicing. Nature. 2013;503:229–234.
  • Soumillon M, Necsulea A, Weier M, et al. Cellular source and mechanisms of high transcriptome complexity in the mammalian testis. Cell Rep. 2013;3:2179–2190.
  • Sharma U, Conine CC, Shea JM, et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science. 2016;351:391–396.
  • Filipowicz W, Bhattacharyya S, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet. 2008;9:102–114.
  • Conine CC, Sun F, Song L, et al. Small RNAs gained during epididymal transit of sperm are essential for embryonic development in mice. Dev Cell. 2018;46(470–480.e3):470–480.e3.
  • Yuan S, Schuster A, Tang C, et al. Sperm-borne miRNAs and endo-siRNAs are important for fertilization and preimplantation embryonic development. Development. 2016;143:635–647.
  • Soubry A. Epigenetics as a driver of developmental origins of health and disease: did we forget the fathers? Bioessays. 2018;40:1.
  • Moholdt T, Hawley JA. Maternal Lifestyle Interventions: targeting Preconception Health. Trends Endocrinol Metab. 2020;S1043-2760(20):30058.
  • Dupont C, Kappeler L, Saget S, et al. Role of miRNA in the transmission of metabolic diseases associated with paternal diet-induced obesity. Front Genet. 2019;10:337.
  • Rodgers AB, Morgan CP, Leu NA, et al. Transgenerational epigenetic programming via sperm microRNA recapitulates effects of paternal stress. Proc Natl Acad Sci U S A. 2015;112(44):13699–13704.
  • Tyebji S, Hannan AJ, Tonkin CJ. Pathogenic infection in male mice changes sperm small RNA profiles and transgenerationally alters offspring behavior. Cell Rep. 2020;31(4):107573.
  • Chan JC, Morgan CP, Adrian LN, et al. Reproductive tract extracellular vesicles are sufficient to transmit intergenerational stress and program neurodevelopment. Nat Commun. 2020;11:1499.
  • Manku G, Culty M. Mammalian gonocyte and spermatogonia differentiation: recent advances and remaining challenges. Reproduction. 2015;149:R139–157.
  • Gan H, Cai T, Lin X, et al. Integrative proteomic and transcriptomic analyses reveal multiple post-transcriptional regulatory mechanisms of mouse spermatogenesis. Mol Cell Proteomics. 2013;12:1144–1157.
  • Liao J, Suen HC, Luk ACS, et al. Transcriptomic and epigenomic profiling of young and aged spermatogonial stem cells reveals molecular targets regulating differentiation. PLoS Genet. 2021;17.
  • Liu W, Wang F, Xu Q, et al. BCAS2 is involved in alternative mRNA splicing in spermatogonia and the transition to meiosis. Nat Commun. 2017;8:14182.
  • Saeidi S, Shapouri F, de Iongh RU, et al. Esrp1 is a marker of mouse fetal germ cells and differentially expressed during spermatogenesis. PLoS ONE. 2018;13:e0190925.
  • Wang X, Lu Z, Gomez A, et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature. 2014;505:117–120.
  • Wang X, Zhao BS, Roundtree IA, et al. N(6)-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161:1388–1399.
  • Xiao W, Adhikari S, Dahal U, et.al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol Cell. 2016;61:507–519.
  • Liu J, Yue Y, Han D, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10:93–95.
  • Lin Z, Hsu PJ, Xing X, et al. Mettl3-/Mettl14-mediated mRNA N6-methyladenosine modulates murine spermatogenesis. Cell Res. 2017;27:1216–1230.
  • Xu K, Yang Y, Feng GH, et al. Mettl3-mediated m6A regulates spermatogonial differentiation and meiosis initiation. Cell Res. 2017;27:1100–1114.
  • Zhang L, Tang J, Haines CJ, et al. c-kit expression profile and regulatory factors during spermatogonial stem cell differentiation. BMC Dev Biol. 2013;13:38.
  • Rossi P, Marziali G, Albanesi C, et al. A novel c-Kit transcript, potentially encoding a truncated receptor, originates within a kit gene intron in mouse spermatids. Dev Biol. 1992;152:203–207.
  • Melino G, De Laurenzi V, Vousden KH. p73: friend or foe in tumorigenesis. Nat Rev Cancer. 2002;2:605–615.
  • Holembowski L, Kramer D, Riedel D, et al. TAp73 is essential for germ cell adhesion and maturation in testis. J Cell Biol. 2014;204:1173–1190.
  • Inoue S, Tomasini R, Rufini A, et al. TAp73 is required for spermatogenesis and the maintenance of male fertility. Proc Natl Acad Sci U S A. 2014;111:1843–1848.
  • Fu K, Tian S, Tan H, et al. Biological and RNA regulatory function of MOV10 in mammalian germ cells. BMC Biol. 2019;17:39.
  • Legrand JMD, Chan AL, La HM, et al. DDX5 plays essential transcriptional and post-transcriptional roles in the maintenance and function of spermatogonia. Nat Commun. 2019;10:2278.
  • Vourekas A, Zheng K, Fu Q, et al. The RNA helicase MOV10L1 binds piRNA precursors to initiate piRNA processing. Genes Dev. 2015;29:617–629.
  • Cheng CY, Mruk DD. Spermatogenesis, Mammals. In: Maloy S, Hughes K, editors. Brenner’s encyclopedia of genetics. 2nd ed. San Diego: Academic Press; 2013. p. 525–528.
  • Schmid R, Grellscheid SN, Ehrmann I, et al. The splicing landscape is globally reprogrammed during male meiosis. Nucleic Acids Res. 2013;41:10170–10184.
  • Jenkins HT, Malkova B, Edwards TA. Kinked β-strands mediate high-affinity recognition of mRNA targets by the germ-cell regulator DAZL. Proc Natl Acad Sci U S A. 2011;108:18266–18271.
  • Reynolds N, Collier B, Bingham V, et al. Translation of the synaptonemal complex component Sycp3 is enhanced in vivo by the germ cell specific regulator Dazl. RNA. 2007;13:974–981.
  • Reynolds N, Collier B, Maratou K, et al. Dazl binds in vivo to specific transcripts and can regulate the pre-meiotic translation of Mvh in germ cells. Hum Mol Genet. 2005;14:3899–3909.
  • Kauppi L, Barchi M, Baudat F, et al. Distinct properties of the XY pseudoautosomal region crucial for male meiosis. Science. 2011;331(6019):916–920.
  • Carofiglio F, Inagaki A, de Vries S, et al. SPO11-independent DNA repair foci and their role in meiotic silencing. PLoS Genet. 2013;9(6):e1003538.
  • Cesari E, Loiarro M, Naro C, et al. Combinatorial control of Spo11 alternative splicing by modulation of RNA polymerase II dynamics and splicing factor recruitment during meiosis. Cell Death Dis. 2020;11:240.
  • Bao J, Tang C, Li J, et al. RNA-binding protein 9 is involved in alternative splicing and is critical for male germ cell development and male fertility. PLoS Genet. 2014;10:e1004825.
  • Puverel S, Barrick C, Dolci S, et al. RanBPM is essential for mouse spermatogenesis and oogenesis. Development. 2011;138:2511–2521.
  • Lemay JF, Lemieux C, St-André O, et al. Crossing the borders: poly(A)-binding proteins working on both sides of the fence. RNA Biol. 2011;7:291–295.
  • Yang J, Zhang Z, Zhang Y, et al. CLOCK interacts with RANBP9 and is involved in alternative splicing in spermatogenesis. Gene. 2018;642:199–204.
  • Paronetto MP, Messina V, Bianchi E, et al. Sam68 regulates translation of target mRNAs in male germ cells, necessary for mouse spermatogenesis. J Cell Biol. 2009;185:235–249.
  • Paronetto MP, Messina V, Barchi M, et al. Sam68 marks the transcriptionally active stages of spermatogenesis and modulates alternative splicing in male germ cells. Nucleic Acids Res. 2011;39:4961–4974.
  • Elliott DJ, Grellscheid SN. Alternative RNA splicing regulation in the testis. Reproduction. 2006;132:811–819.
  • Venables JP, Bourgeois CF, Dalgliesh C, et al. Up-regulation of the ubiquitous alternative splicing factor Tra2beta causes inclusion of a germ cell-specific exon. Hum Mol Genet. 2005;14:2289–2303.
  • Liu Y, Bourgeois CF, Pang S, et al. The germ cell nuclear proteins hnRNP G-T and RBMY activate a testis-specific exon. PLoS Genet. 2009;5:e1000707.
  • Dreumont N, Bourgeois CF, Lejeune F, et al. Human RBMY regulates germline-specific splicing events by modulating the function of the serine/arginine-rich proteins 9G8 and Tra2-beta. J Cell Sci. 2010;123:40–50.
  • Pedrotti S, Busà R, Compagnucci C, et al. The RNA recognition motif protein RBM11 is a novel tissue-specific splicing regulator. Nucleic Acids Res. 2012;40:1021–1032.
  • Di Giammartino DC, Nishida K, Manley JL. Mechanisms and consequences of alternative polyadenylation. Mol Cell. 2011;43:853–866.
  • Jia J, Yao P, Arif A, et al. Regulation and dysregulation of 3′ UTR-mediated translational control. Curr Opin Genet Dev. 2013;23:29–34.
  • MacDonald CC, McMahon KW. Tissue-specific mechanisms of alternative polyadenylation: testis, brain, and beyond. Wiley Interdiscip Rev RNA. 2010;1:494–501.
  • Zhang Y, Tang C, Yu T, et al. MicroRNAs control mRNA fate by compartmentalization based on 3ʹ UTR length in male germ cells. Genome Biol. 2017;18:105.
  • Monteuuis G, Wong JJL, Bailey CG, et al. The changing paradigm of intron retention: regulation, ramifications and recipes. Nucleic Acids Res. 2019;47:11497–11513.
  • Wong JJ, Au AY, Ritchie W, et al. Intron retention in mRNA: no longer nonsense: known and putative roles of intron retention in normal and disease biology. Bioessays. 2016;38:41–49.
  • Naro C, Jolly A, Di Persio S, et al. An orchestrated intron retention program in meiosis controls timely usage of transcripts during germ cell differentiation. Dev Cell. 2017;41:82–93.
  • Izumikawa K, Yoshikawa H, Ishikawa H, et al. Chtop (Chromatin target of Prmt1) auto-regulates its expression level via intron retention and nonsense-mediated decay of its own mRNA. Nucleic Acids Res. 2016;44:9847–9859.
  • Ullrich S, Guigó R. Dynamic changes in intron retention are tightly associated with regulation of splicing factors and proliferative activity during B-cell development. Nucleic Acids Res. 2020;48:1327–1340.
  • Wong JJ, Ritchie W, Ebner OA, et al. Orchestrated intron retention regulates normal granulocyte differentiation. Cell. 2013;154:583–595.
  • Yao J, Ding D, Li X, et al. Prevalent intron retention fine-tunes gene expression and contributes to cellular senescence. Aging Cell. 2020;19:e13276.
  • Paronetto MP, Sette C. Role of RNA-binding proteins in mammalian spermatogenesis. Int J Androl. 2010;33:2–12.
  • Gaucher J, Boussouar F, Montellier E, et al. Bromodomain-dependent stage-specific male genome programming by Brdt. EMBO J. 2012;31:3809–3820.
  • Berkovits BD, Wang L, Guarnieri P, et al. The testis-specific double bromodomain-containing protein BRDT forms a complex with multiple spliceosome components and is required for mRNA splicing and 3ʹ-UTR truncation in round spermatids. Nucleic Acids Res. 2012;40:7162–7175.
  • Manterola M, Brown TM, Oh MY, et al. BRDT is an essential epigenetic regulator for proper chromatin organization, silencing of sex chromosomes and crossover formation in male meiosis. PLoS Genet. 2018;14:e1007209.
  • Zheng G, Dahl JA, Niu Y, et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol Cell. 2013;49:18–29.
  • Tang C, Klukovich R, Peng H, et al. ALKBH5-dependent m6A demethylation controls splicing and stability of long 3′-UTR mRNAs in male germ cells. Proc Natl Acad Sci U S A. 2018;115:E325–E333.
  • Hsu PJ, Zhu Y, Ma H, et al. Ythdc2 is an N6-methyladenosine binding protein that regulates mammalian spermatogenesis. Cell Res. 2017;27:1115–1127.
  • Wojtas MN, Pandey RR, Mendel M, et al. Regulation of m6A transcripts by the 3ʹ→5ʹ RNA helicase YTHDC2 is essential for a successful meiotic program in the mammalian germline. Mol Cell. 2017;68:374–387.e12.
  • Abby E, Tourpin S, Ribeiro J, et al. Implementation of meiosis prophase I programme requires a conserved retinoid-independent stabilizer of meiotic transcripts. Nat Commun. 2016;7:10324.
  • Bailey AS, Batista PJ, Gold RS, et al. The conserved RNA helicase YTHDC2 regulates the transition from proliferation to differentiation in the germline. Elife. 2017;6:e26116.
  • Hannigan MM, Zagore LL, Licatalosi DD. Ptbp2 controls an alternative splicing network required for cell communication during spermatogenesis. Cell Rep. 2017;19:2598–2612.
  • Zagore LL, Grabinski SE, Sweet TJ, et al. RNA binding protein Ptbp2 is essential for male germ cell development. Mol Cell Biol. 2015;35:4030–4042.
  • Pivot-Pajot C, Caron C, Govin J, et al. Acetylation-dependent chromatin reorganization by BRDT, a testis-specific bromodomain-containing protein. Mol Cell Biol. 2003;23:5354–5365.
  • Rathke C, Baarends WM, Awe S, et al. Chromatin dynamics during spermiogenesis. Biochim Biophys Acta. 2014;1839:155–168.
  • Dhar S, Thota A, Rao MR. Insights into role of bromodomain, testis-specific (Brdt) in acetylated histone H4-dependent chromatin remodeling in mammalian spermiogenesis. J Biol Chem. 2012;287:6387–6405.
  • Morinière J, Rousseaux S, Steuerwald U, et al. Cooperative binding of two acetylation marks on a histone tail by a single bromodomain. Nature. 2009;461:664–668.
  • Luco RF, Pan Q, Tominaga K, et al. Regulation of alternative splicing by histone modifications. Science. 2010;327:996–1000.
  • Iwamori N, Tominaga K, Sato T, et al. MRG15 is required for pre-mRNA splicing and spermatogenesis. Proc Natl Acad Sci U S A. 2016;113:E5408–15.
  • O’Bryan MK, Clark BJ, McLaughlin EA, et al. RBM5 is a male germ cell splicing factor and is required for spermatid differentiation and male fertility. PLoS Genet. 2013;9:e1003628.
  • Bonnal S, Martinez C, Forch P, et al. RBM5/Luca-15/H37 regulates Fas alternative splice site pairing after exon definition. Mol Cell. 2008;32:81–95.
  • Fushimi K, Ray P, Kar A, et al. Up-regulation of the proapoptotic caspase 2 splicing isoform by a candidate tumor suppressor, RBM5. Proc Natl Acad Sci U S A. 2008;105:15708–15713.
  • Mourão A, Bonnal S, Soni K, et al. Structural basis for the recognition of spliceosomal SmN/B/B’ proteins by the RBM5 OCRE domain in splicing regulation. Elife. 2016;5:e14707.
  • Baba T, Niida Y, Michikawa Y, et al. An acrosomal protein, sp32, in mammalian sperm is a binding protein specific for two proacrosins and an acrosin intermediate. J Biol Chem. 1994;269:10133–10140.
  • Kanemori Y, Ryu JH, Sudo M, et al. Two functional forms of ACRBP/sp32 are produced by premRNA alternative splicing in the mouse. Biol Reprod. 2013;88:105.
  • Kanemori Y, Koga Y, Sudo M, et al. Biogenesis of sperm acrosome is regulated by pre-mRNA alternative splicing of Acrbp in the mouse. Proc Natl Acad Sci U S A. 2016;113:E3696–3705.
  • Fang EF, Scheibye-Knudsen M, Chua KF, et al. Nuclear DNA damage signalling to mitochondria in ageing. Nat Rev Mol Cell Biol. 2016;17(5):308–321.
  • Tatone C, Di Emidio G, Barbonetti A, et al. Sirtuins in gamete biology and reproductive physiology: emerging roles and therapeutic potential in female and male infertility. Hum Reprod Update. 2018;24:267–289.
  • Michishita E, Park JY, Burneskis JM, et al. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol Biol Cell. 2005;16:4623–4635.
  • Bell EL, Nagamori I, Williams EO, et al. SirT1 is required in the male germ cell for differentiation and fecundity in mice. Development. 2014;141:3495–3504.
  • Di Sante G, Wang L, Wang C, et al. Sirt1-defificient mice have hypogonadotropic hypogonadism due to defective GnRH neuronal migration. Mol Endocrinol. 2015;29:200–212.
  • Jiao D, Zhang H, Jiang Z, et al. MicroRNA-34a targets sirtuin 1 and leads to diabetes-induced testicular apoptotic cell death. J Mol Med. 2018;96:939–949.
  • Helmy HS, Senousy MA, El-Sahar AE, et al. Aberrations of miR-126-3p, miR-181a and Sirtuin1 Network Mediate Di-(2-ethylhexyl) phthalate-induced Testicular Damage in Rats: the Protective Role of Hesperidin. Toxicology. 2020;152406. 10.1016/j.tox.2020.152406
  • Chalkiadaki A, Guarente L. The multifaceted functions of sirtuins in cancer. Nat Rev Cancer. 2015;15:608–624.
  • Cheng A, Yang Y, Zhou Y, et al. Mitochondrial SIRT3 mediates adaptive responses of neurons to exercise and metabolic and excitatory challenges. Cell Metab. 2016;23:128–142.
  • North BJ, Rosenberg MA, Jeganathan KB, et al. SIRT2 induces the checkpoint kinase BubR1 to increase lifespan. EMBO J. 2014;33:1438–1453.
  • Kanfi Y, Naiman S, Amir G, et al. The sirtuin SIRT6 regulates lifespan in male mice. Nature. 2012;483:218–221.
  • Mostoslavsky R, Chua KF, Lombard DB, et al. Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell. 2006;124:315–329.
  • Ryu D, Jo YS, Lo Sasso G, et al. A SIRT7-dependent acetylation switch of GABPβ1 controls mitochondrial function. Cell Metab. 2014;20:856–869.
  • Dai X, Yan X, Wintergerst KA, et al. Nrf2: redox and metabolic regulator of stem cell state and function. Trends Mol Med. 2020;26:185–200.
  • Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39:199–218.
  • Silva-Palacios A, Ostolga-Chavarría M, Zazueta C, et al. Nrf2: molecular and epigenetic regulation during aging. Ageing Res Rev. 2018;47:31–40.
  • Wang H, Liu K, Geng M, et al. RXRα inhibits the NRF2-ARE signaling pathway through a direct interaction with the Neh7 domain of NRF2. Cancer Res. 2013;73:3097–3108.
  • Moldogazieva NT, Lutsenko SV, Terentiev AA. Reactive oxygen and nitrogen species-induced protein modifications: implication in carcinogenesis and anticancer therapy. Cancer Res. 2018;78(21):6040–6047.
  • Azzimato V, Jager J, Chen P, et al. Liver macrophages inhibit the endogenous antioxidant response in obesity-associated insulin resistance. Sci Transl Med. 2020;12:eaaw9709.
  • Qin Z, Zhu K, Xue J, et al. Zinc-induced protective effect for testicular ischemia-reperfusion injury by promoting antioxidation via microRNA-101-3p/Nrf2 pathway. Aging (Albany NY). 2019;11:9295–9309.
  • Zhao XJ, Yu HW, Yang YZ, et al., Polydatin prevents fructose-induced liver inflammation and lipid deposition through increasing miR-200a to regulate Keap1/Nrf2 pathway. Redox Biol Redox Biol. 2018; 18:124–137.
  • Poganik JR, Long MJC, Disare MT, et al. Post-transcriptional regulation of Nrf2-mRNA by the mRNA-binding proteins HuR and AUF1. FASEB J. 2019;33:14636–14652.
  • Sánchez-Martín P, Komatsu M. p62/SQSTM1-steering the cell through health and disease. J Cell Sci. 2018;131(21):jcs222836.
  • Yang C, Yao C, Tian R, et al. miR-202-3p regulates sertoli cell proliferation, synthesis function, and apoptosis by targeting LRP6 and cyclin D1 of Wnt/β-catenin signaling. Mol Ther Nucleic Acids. 2019;14:1–19.
  • Leung L, Kwong M, Hou S, et al. Deficiency of the Nrf1 and Nrf2 transcription factors results in early embryonic lethality and severe oxidative stress. J Biol Chem. 2003;278(48):48021–48029.
  • Wakabayashi N, Itoh K, Wakabayashi J, et al. Keap1-null mutation leads to postnatal lethality due to constitutive Nrf2 activation. Nat Genet. 2003;35:238–245.
  • Nakamura BN, Lawson G, Chan JY, et al. Knockout of the transcription factor NRF2 disrupts spermatogenesis in an age-dependent manner. Free Radic Biol Med. 2010;49:1368–1379.
  • Chen K, Mai Z, Zhou Y, et al. Low NRF2 mRNA expression in spermatozoa from men with low sperm motility. Tohoku J Exp Med. 2012;228(3):259–266.
  • Yu B, Lin H, Yang L, et al. Genetic variation in the Nrf2 promoter associates with defective spermatogenesis in humans. J Mol Med (Berl). 2012;90:1333–1342.
  • López P, Castro A, Flórez M, et al. miR-155 and miR-122 Expression of Spermatozoa in Obese Subjects. Front Genet. 2018;9:175.
  • Yehuda R, Lehrner A. Intergenerational transmission of trauma effects: putative role of epigenetic mechanisms. World Psychiatry. 2018;17:243–257.
  • Fullston T, Ohlsson-Teague EM, Print CG, et al. Sperm microRNA content is altered in a mouse model of male obesity, but the same suite of micrornas are not altered in offspring’s sperm. PLoS One. 2016;11:e0166076.
  • Cropley JE, Eaton SA, Aiken A, et al. Male-lineage transmission of an acquired metabolic phenotype induced by grand-paternal obesity. Mol Metab. 2016;5:699–708.
  • de Castro Barbosa T, Ingerslev LR, Alm PS, et al. High-fat diet reprograms the epigenome of rat spermatozoa and transgenerationally affects metabolism of the offspring. Mol Metab. 2015;5:184–197.
  • Grandjean V, Fourré S, De Abreu DA, et al. RNA-mediated paternal heredity of diet-induced obesity and metabolic disorders. Sci Rep. 2015;5:18193.
  • Murashov AK, Pak ES, Koury M; Murashov AK, Pak ES, Koury M, Ajmera A, Jeyakumar M, Williams O, Ding J, Walters D, Neufer PD. Paternal long-term exercise programs offspring for low energy expenditure and increased risk for obesity in mice. FASEB J. 2016;30:775–784.
  • Carone BR, Fauquier L, Habib N, et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell. 2010;143:1084–1096.
  • Dickson DA, Paulus JK, Mensah V, et al. Reduced levels of miRNAs 449 and 34 in sperm of mice and men exposed to early life stress. Transl Psychiatry. 2018;8:101.
  • Cheeseman K, Weitzman JB. Host-parasite interactions: an intimate epigenetic relationship. Cell Microbiol. 2015;17(8):1121–1132.
  • Pellati D, Mylonakis I, Bertoloni G, et al. Genital tract infections and infertility. Eur J Obstet Gynecol Reprod Biol. 2008;140:3–11.
  • Knuesel I, Chicha L, Britschgi M, et al. Maternal immune activation and abnormal brain development across CNS disorders. Nat Rev Neurol. 2014;10:643–660.
  • Melamed Z, Levy A, Ashwal-Fluss R, et al. Alternative splicing regulates biogenesis of miRNAs located across exon-intron junctions. Mol Cell. 2013;50:869–881.
  • Levine DC, Hong H, Weidemann BJ, et al. NAD controls circadian reprogramming through PER2 nuclear translocation to counter aging. Mol Cell. 2020;78:835–849.
  • Timmers S, Konings E, Bilet L, et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011;14:612–622.
  • Saito Y, Yako T, Otsu W, et al. A triterpenoid Nrf2 activator, RS9, promotes LC3-associated phagocytosis of photoreceptor outer segments in a p62-independent manner. Free Radic Biol Med. 2020;152:235–247.
  • Panneerdoss S, Viswanadhapalli S, Abdelfattah N, et al. Cross-talk between miR-471-5p and autophagy component proteins regulates LC3-associated phagocytosis (LAP) of apoptotic germ cells. Nat Commun. 2017;8:598.
  • Chen Q, Yan M, Cao Z, et al. Sperm tsRNAs contribute to intergenerational inheritance of an acquired metabolic disorder. Science. 2016;351(6271):397–400.
  • Kazachenka A, Bertozzi TM, Sjoberg-Herrera MK, et al. Identification, characterization, and heritability of murine metastable epialleles: implications for non-genetic inheritance. Cell. 2018;175(5):1259–1271.e13.
  • Kiani J, Grandjean V, Liebers R, et al. RNA-mediated epigenetic heredity requires the cytosine methyltransferase Dnmt2. PLoS Genet. 2013;9(5):e1003498.
  • Ding N, Zhang X, Zhang XD, et al. Impairment of spermatogenesis and sperm motility by the high-fat diet-induced dysbiosis of gut microbes. Gut. 2020;gutjnl-2019-319127.
  • Qi X, Yun C, Sun L, et al. Gut microbiota-bile acidinterleukin-22 axis orchestrates polycystic ovary syndrome. Nat Med. 2019;25:1225–1233.
  • Wang X, Li Y, Chen W, et al. Transcriptome-wide reprogramming of N6-methyladenosine modification by the mouse microbiome. Cell Res. 2019;29:167–170.
  • Skvortsova K, Iovino N, Bogdanovic O. Functions and mechanisms of epigenetic inheritance in animals. Nat Rev Mol Cell Biol. 2018;19:774–790.
  • Gao H, Wen H, Cao C, et al. Overexpression of MicroRNA-10a in germ cells causes male infertility by targeting Rad51 in mouse and human. Front Physiol. 2019;10:765.
  • Li L, Zhu Y, Chen T, et al. MiR-125b-2 knockout in testis is associated with targeting to the PAP gene, mitochondrial copy number, and impaired sperm quality. Int J Mol Sci. 2019;20:148.
  • Chen J, Cai T, Zheng C, et al. MicroRNA-202 maintains spermatogonial stem cells by inhibiting cell cycle regulators and RNA binding proteins. Nucleic Acids Res. 2017;45:4142–4157.
  • Cui N, Hao G, Zhao Z, et al. MicroRNA-224 regulates self-renewal of mouse spermatogonial stem cellsviatargeting DMRT1. J Cell Mol Med. 2016;20(8):1503–1512.
  • Bao J, Li D, Wang L, et al. MicroRNA-449 and MicroRNA-34b/c function redundantly in murine testes by targeting E2F transcription factor-retinoblastoma Protein (E2F-pRb) pathway. J Biol Chem. 2012;287:21686–21698.
  • Comazzetto S, Di Giacomo M, Rasmussen KD, et al. Oligoasthenoteratozoospermia and infertility in mice deficient for miR-34b/c and miR-449 Loci. PLoS Genet. 2014;10:e1004597.
  • Moritoki Y, Hayashi Y, Mizuno K, et al. Expression profiling of microRNA in cryptorchid testes: miR-135a contributes to the maintenance of spermatogonial stem cells by regulating FoxO1. J Urol. 2014;191:1174–1180.
  • Yang QE, Racicot KE, Kaucher AV, et al. MicroRNAs 221 and 222 regulate the undifferentiated state in mammalian male germ cells. Development. 2012;140(2):280–290.
  • Dai L, Tsai-Morris CH, Sato H, et al. Testis-specific miRNA-469 up-regulated in gonadotropin-regulated testicular RNA helicase (GRTH/DDX25)-null mice silences transition protein 2 and protamine 2 messages at sites within coding region. J Biol Chem. 2011;286:44306–44318.
  • Niu Z, Goodyear SM, Rao S, et al. MicroRNA-21 regulates the self-renewal of mouse spermatogonial stem cells. Proc Nat Acad Sci. 2011;108:12740–12745.
  • Ramaiah M, Tan K, Plank TM, et al. A microRNA cluster in the Fragile-X region expressed during spermatogenesis targets FMR1. EMBO Rep. 2019;20(2):e46566.
  • Yang Y, Willis TL, Button RW, et al. Cytoplasmic DAXX drives SQSTM1/p62 phase condensation to activate Nrf2-mediated stress response. Nat Commun. 2019;10:3759.
  • Zhang L, Ma J, Yang B, et al. Interference with lactate metabolism by mmu-miR-320-3p via negatively regulating GLUT3 signaling in mouse Sertoli cells. Cell Death Dis. 2018;9:10.
  • Xu Y, Wu W, Fan Y, et al. MiR-142-3p inhibits TGF-β3-induced blood-testis barrier impairment by targeting lethal giant larvae homolog 2. Cell Physiol Biochem. 2018;46(1):253–268.
  • Nakagawa T, Nabeshima Y, Yoshida S. Functional Identification of the actual and potential stem cell compartments in mouse spermatogenesis. Dev Cell. 2007;12:195–206.